Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 71
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Med Virol ; 96(1): e29417, 2024 01.
Artículo en Inglés | MEDLINE | ID: mdl-38258345

RESUMEN

The EG.5.1 variant of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) has been prevalent since mid-July 2023 in the United States and China. The variant BA.2.86 has become a major concern because it is 34 mutations away from the parental variant BA.2 and >30 mutations from XBB.1.5. There is an urgent need to evaluate whether the immunity of the population and current vaccines are protective against EG.5.1 and BA.2.86. Based on a cohort of two breakthrough-infected groups, the levels of neutralizing antibodies (NAbs) against different subvariants were measured using pseudovirus-based neutralization assays. XBB.1.5, EG.5.1, and BA.2.86 are comparably immune-evasive from neutralization by the plasma of individuals recovered from BA.5 infection (BA.5-convalescent) or XBB.1.9.2/XBB.1.5 infection following BA.5 infection (BA.5-XBB-convalescent). NAb levels against EG.5.1 and BA.2.86 subvariants remained >120 geometric mean titers (GMTs) in BA.5-XBB-convalescent individuals 2 months postinfection but were <40 GMTs in BA.5-convalescent individuals. Furthermore, the XBB-targeting messenger RNA (mRNA) vaccine RQ3033 induced higher levels of NAbs against XBB.1.5, EG.5.1, and BA.2.86 than against BA.5-XBB infection. The results suggest that BA.2.86 and EG.5.1 are unlikely to cause more severe concerns than the currently circulating XBB subvariants and that the XBB.1.5-targeting mRNA vaccine tested has promising protection against EG.5.1 and BA.2.86.


Asunto(s)
Anticuerpos Neutralizantes , Plasma , Humanos , China , Evasión Inmune , Mutación , ARN Mensajero , SARS-CoV-2/genética
2.
J Med Virol ; 96(3): e29454, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38445768

RESUMEN

Various vaccines have been challenged by SARS-CoV-2 variants. Here, we reported a yeast-derived recombinant bivalent vaccine (Bivalent wild-type [Wt]+De) based on the wt and Delta receptor-binding domain (RBD). Yeast derived RBD proteins based on the wt and Delta mutant were used as the prime vaccine. It was found that, in the presence of aluminium hydroxide (Alum) and unmethylated CpG-oligodeoxynucleotides (CpG) adjuvants, more cross-protective immunity against SARS-CoV-2 prototype and variants were elicited by bivalent vaccine than monovalent wtRBD or Delta RBD. Furthermore, a heterologous boosting strategy consisting of two doses of bivalent vaccines followed by one dose adenovirus vectored vaccine exhibited cross-neutralization capacity and specific T cell responses against Delta and Omicron (BA.1 and BA.4/5) variants in mice, superior to a homologous vaccination strategy. This study suggested that heterologous prime-boost vaccination with yeast-derived bivalent protein vaccine could be a potential approach to address the challenge of emerging variants.


Asunto(s)
COVID-19 , Vacunas , Animales , Ratones , Vacunas Combinadas , Proteínas Fúngicas , Saccharomyces cerevisiae/genética , COVID-19/prevención & control , SARS-CoV-2 , Vacunación
3.
J Med Virol ; 96(3): e29544, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38511577

RESUMEN

The impact of SARS-CoV-2 infection shortly after vaccination on vaccine-induced immunity is unknown, which is also one of the concerns for some vaccinees during the pandemic. Here, based on a cohort of individuals who encountered BA.5 infection within 8 days after receiving the fourth dose of a bivalent mRNA vaccine, preceded by three doses of inactivated vaccines, we show that booster mRNA vaccination provided 48% protection efficacy against symptomatic infections. At Day 7 postvaccination, the level of neutralizing antibodies (Nabs) against WT and BA.5 strains in the uninfected group trended higher than those in the symptomatic infection group. Moreover, there were greater variations in Nabs levels and a significant decrease in virus-specific CD4+ T cell response observed in the symptomatic infection group. However, symptomatic BA.5 infection significantly increased Nab levels against XBB.1.9.1 and BA.5 (symptomatic > asymptomatic > uninfected group) at Day 10 and resulted in a more gradual decrease in Nabs against BA.5 compared to the uninfected group at Day 90. Our data suggest that BA.5 infection might hinder the early generation of Nabs and the recall of the CD4+ T cell response but strengthens the Nab and virus-specific T cell response in the later phase. Our data confirmed that infection can enhance host immunity regardless of the short interval between vaccination and infection and alleviate concerns about infections shortly after vaccination, which provides valuable guidance for developing future vaccine administration strategies.


Asunto(s)
Anticuerpos Neutralizantes , Vacunación , Humanos , Inmunización Secundaria , ARN Mensajero/genética , Vacunas Combinadas , Anticuerpos Antivirales
4.
J Immunol ; 209(10): 1832-1836, 2022 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-36426954

RESUMEN

In this study, we investigated how pre-existing Ab immunity to influenza virus established from prior immunizations affects the development of CD8+ T cell responses evoked after vaccination with a live attenuated vaccine. Using a mouse model and a panel of live attenuated influenza virus vaccine candidates (cold adapted and single cycle), we show that pre-existing influenza-specific Abs directed against the vaccine backbone attenuate the size and quality of the vaccine-induced CD8+ T cell response. Importantly, we show that increasing the vaccine dose can overcome this impediment, resulting in improved vaccine-induced circulating and tissue-resident memory CD8+ T cell responses, which were protective against heterologous influenza challenge. Thus, the reduced size and quality of the T cell response elicited by a live attenuated influenza virus vaccine imparted by the influenza-specific Ab landscape of the vaccinee can be overcome by increasing vaccine dose.


Asunto(s)
Vacunas contra la Influenza , Gripe Humana , Humanos , Vacunas Atenuadas , Inmunidad Humoral , Linfocitos T CD8-positivos
5.
J Med Virol ; 95(1): e28237, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36258299

RESUMEN

With the continuation of the coronavirus disease 2019 pandemic and the emergence of new severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) variants, the control of the spread of the virus remains urgent. Various animals, including cats, ferrets, hamsters, nonhuman primates, minks, tree shrews, fruit bats, and rabbits, are susceptible to SARS-CoV-2 infection naturally or experimentally. Therefore, to avoid animals from becoming mixing vessels of the virus, vaccination of animals should be considered. In the present study, we report the establishment of an efficient and stable system using Newcastle disease virus (NDV) as a vector to express SARS-CoV-2 spike protein/subunit for the rapid generation of vaccines against SARS-CoV-2 in animals. Our data showed that the S and S1 protein was sufficiently expressed in rNDV-S and rNDV-S1-infected cells, respectively. The S protein was incorporated into and displayed on the surface of rNDV-S viral particles. Intramuscular immunization with rNDV-S was found to induce the highest level of binding and neutralizing antibodies, as well as strong S-specific T-cell response in mice. Intranasal immunization with rNDV-S1 provoked a robust T-cell response but barely any detectable antibodies. Overall, the NDV-vectored vaccine candidates were able to induce profound humoral and cellular immunity, which will provide a good system for developing vaccines targeting both T-cell and antibody responses.


Asunto(s)
COVID-19 , Vacunas Virales , Animales , Ratones , Humanos , Conejos , Vacunas contra la COVID-19 , Virus de la Enfermedad de Newcastle/genética , SARS-CoV-2 , COVID-19/prevención & control , Hurones/metabolismo , Glicoproteína de la Espiga del Coronavirus/genética , Anticuerpos Neutralizantes/metabolismo , Anticuerpos Antivirales/metabolismo , Vacunas Virales/genética
6.
Immunopharmacol Immunotoxicol ; 45(1): 16-25, 2023 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-35850595

RESUMEN

BACKGROUND: The first-line anti-rheumatic drug methotrexate (MTX) is used in the combination. Because of the unpredictable adverse reactions, optimization of relevant regimens is necessary and meaningful. This study aimed to study the possible interaction between Securidaca inappendiculate Hassk. Derived xanthones and MTX. METHODS: We established adjuvant-induced arthritis (AIA) model, which was treated with MTX and MTX + xanthone-rich fraction (XRF). The clinical efficacy was evaluated by histopathological examination, and LC-MS was used to monitor the blood concentration of MTX. Western blotting and immunohistochemistry were used to detect protein expression. In vitro, we assessed the activity of related transporters by cellular uptake assay based on HEK-293T cells. RESULTS: Compared with MTX-treated rats, inflammation in the immunized rats in the MTX + XRF group was obvious, indicating that XRF antagonized the anti-rheumatic effect of MTX. Meanwhile, XRF reduced liver and kidney injuries caused by MTX in addition to MTX. Results from immunohistochemical and nappendiculat assays suggested that XRF may reduce uptake of MTX by down-regulating reduced folate carrier 1 (RFC1). CONCLUSION: This study indicated that XRF could reduce the plasma concentration of MTX by inhibiting the expression of RFC1, antagonize the therapeutic effect of MTX on AIA rats, and reduce its oral bioavailability. The combination of S. inappendiculate and MTX should be further optimized to achieve the goal of increasing efficiency and reducing toxicity.


Asunto(s)
Antirreumáticos , Securidaca , Xantonas , Ratas , Animales , Metotrexato/farmacología , Securidaca/metabolismo , Proteína Portadora de Folato Reducido , Antirreumáticos/farmacología , Antirreumáticos/uso terapéutico , Xantonas/farmacología
7.
Immunol Cell Biol ; 100(7): 547-561, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35514192

RESUMEN

Mucosal-associated invariant T (MAIT) cells are a major subset of innate-like T cells mediating protection against bacterial infection through recognition of microbial metabolites derived from riboflavin biosynthesis. Mouse MAIT cells egress from the thymus as two main subpopulations with distinct functions, namely, T-bet-expressing MAIT1 and RORγt-expressing MAIT17 cells. Previously, we reported that inducible T-cell costimulator and interleukin (IL)-23 provide essential signals for optimal MHC-related protein 1 (MR1)-dependent activation and expansion of MAIT17 cells in vivo. Here, in a model of tularemia, in which MAIT1 responses predominate, we demonstrate that IL-12 and IL-23 promote MAIT1 cell expansion during acute infection and that IL-12 is indispensable for MAIT1 phenotype and function. Furthermore, we showed that the bias toward MAIT1 or MAIT17 responses we observed during different bacterial infections was determined and modulated by the balance between IL-12 and IL-23 and that these responses could be recapitulated by cytokine coadministration with antigen. Our results indicate a potential for tailored immunotherapeutic interventions via MAIT cell manipulation.


Asunto(s)
Infecciones Bacterianas , Células T Invariantes Asociadas a Mucosa , Animales , Citocinas , Antígenos de Histocompatibilidad Clase I/metabolismo , Interleucina-12 , Interleucina-23 , Ratones
8.
J Virol ; 94(15)2020 07 16.
Artículo en Inglés | MEDLINE | ID: mdl-32434886

RESUMEN

Middle East respiratory syndrome coronavirus (MERS-CoV) causes severe acute respiratory disease in humans. MERS-CoV strains from early epidemic clade A and contemporary epidemic clade B have not been phenotypically characterized to compare their abilities to infect cells and mice. We isolated the clade B MERS-CoV ChinaGD01 strain from a patient infected during the South Korean MERS outbreak in 2015 and compared the phylogenetics and pathogenicity of MERS-CoV EMC/2012 (clade A) and ChinaGD01 (clade B) in vitro and in vivo Genome alignment analysis showed that most clade-specific mutations occurred in the orf1ab gene, including mutations that were predicted to be potential glycosylation sites. Minor differences in viral growth but no significant differences in plaque size or sensitivity to beta interferon (IFN-ß) were detected between these two viruses in vitro ChinaGD01 virus infection induced more weight loss and inflammatory cytokine production in human DPP4-transduced mice. Viral titers were higher in the lungs of ChinaGD01-infected mice than with EMC/2012 infection. Decreased virus-specific CD4+ and CD8+ T cell numbers were detected in the lungs of ChinaGD01-infected mice. In conclusion, MERS-CoV evolution induced changes to reshape its pathogenicity and virulence in vitro and in vivo and to evade adaptive immune response to hinder viral clearance.IMPORTANCE MERS-CoV is an important emerging pathogen and causes severe respiratory infection in humans. MERS-CoV strains from early epidemic clade A and contemporary epidemic clade B have not been phenotypically characterized to compare their abilities to infect cells and mice. In this study, we showed that a clade B virus ChinaGD01 strain caused more severe disease in mice, with delayed viral clearance, increased inflammatory cytokines, and decreased antiviral T cell responses, than the early clade A virus EMC/2012. Given the differences in pathogenicity of different clades of MERS-CoV, periodic assessment of currently circulating MERS-CoV is needed to monitor potential severity of zoonotic disease.


Asunto(s)
Infecciones por Coronavirus/virología , Genotipo , Interacciones Huésped-Patógeno , Coronavirus del Síndrome Respiratorio de Oriente Medio/fisiología , Adulto , Animales , Modelos Animales de Enfermedad , Genoma Viral , Interacciones Huésped-Patógeno/inmunología , Humanos , Interferón Tipo I/farmacología , Masculino , Ratones , Coronavirus del Síndrome Respiratorio de Oriente Medio/clasificación , Coronavirus del Síndrome Respiratorio de Oriente Medio/aislamiento & purificación , Coronavirus del Síndrome Respiratorio de Oriente Medio/patogenicidad , Filogenia , ARN Viral , Linfocitos T/inmunología , Linfocitos T/metabolismo , Virulencia , Replicación Viral/efectos de los fármacos , Replicación Viral/genética , Secuenciación Completa del Genoma
9.
Proc Natl Acad Sci U S A ; 114(20): 5225-5230, 2017 05 16.
Artículo en Inglés | MEDLINE | ID: mdl-28461487

RESUMEN

The lymphoid tissue that drains the upper respiratory tract represents an important induction site for cytotoxic T lymphocyte (CTL) immunity to airborne pathogens and intranasal vaccines. Here, we investigated the role of the nasal-associated lymphoid tissues (NALTs), which are mucosal-associated lymphoid organs embedded in the submucosa of the nasal passage, in the initial priming and recall expansion of CD8+ T cells following an upper respiratory tract infection with a pathogenic influenza virus and immunization with a live attenuated influenza virus vaccine. Whereas NALTs served as the induction site for the recall expansion of memory CD8+ T cells following influenza virus infection or vaccination, they failed to support activation of naïve CD8+ T cells. Strikingly, NALTs, unlike other lymphoid tissues, were not routinely surveyed during the steady state by circulating T cells. The selective recruitment of memory T cells into these lymphoid structures occurred in response to infection-induced elevation of the chemokine CXCL10, which attracted CXCR3+ memory CD8+ T cells. These results have significant implications for intranasal vaccines, which deliver antigen to mucosal-associated lymphoid tissue and aim to elicit protective CTL-mediated immunity.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Inmunidad Mucosa/inmunología , Linfocitos T Citotóxicos/inmunología , Administración Intranasal , Animales , Inmunización , Virus de la Influenza A/inmunología , Vacunas contra la Influenza/inmunología , Ganglios Linfáticos/fisiología , Tejido Linfoide/metabolismo , Ratones , Ratones Endogámicos C57BL , Mucosa Nasal/metabolismo , Mucosa Nasal/fisiología , Infecciones por Orthomyxoviridae/inmunología , Infecciones del Sistema Respiratorio , Vacunación
11.
Proc Natl Acad Sci U S A ; 113(36): 10133-8, 2016 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-27543331

RESUMEN

Mucosal-associated invariant T (MAIT) cells are innate-like T lymphocytes known to elicit potent immunity to a broad range of bacteria, mainly via the rapid production of inflammatory cytokines. Whether MAIT cells contribute to antiviral immunity is less clear. Here we asked whether MAIT cells produce cytokines/chemokines during severe human influenza virus infection. Our analysis in patients hospitalized with avian H7N9 influenza pneumonia showed that individuals who recovered had higher numbers of CD161(+)Vα7.2(+) MAIT cells in peripheral blood compared with those who succumbed, suggesting a possible protective role for this lymphocyte population. To understand the mechanism underlying MAIT cell activation during influenza, we cocultured influenza A virus (IAV)-infected human lung epithelial cells (A549) and human peripheral blood mononuclear cells in vitro, then assayed them by intracellular cytokine staining. Comparison of influenza-induced MAIT cell activation with the profile for natural killer cells (CD56(+)CD3(-)) showed robust up-regulation of IFNγ for both cell populations and granzyme B in MAIT cells, although the individual responses varied among healthy donors. However, in contrast to the requirement for cell-associated factors to promote NK cell activation, the induction of MAIT cell cytokine production was dependent on IL-18 (but not IL-12) production by IAV-exposed CD14(+) monocytes. Overall, this evidence for IAV activation via an indirect, IL-18-dependent mechanism indicates that MAIT cells are protective in influenza, and also possibly in any human disease process in which inflammation and IL-18 production occur.


Asunto(s)
Inmunidad Mucosa , Subtipo H7N9 del Virus de la Influenza A/patogenicidad , Gripe Humana/inmunología , Interleucina-18/inmunología , Células T Invariantes Asociadas a Mucosa/inmunología , Neumonía Viral/inmunología , Células A549 , Comunicación Celular/inmunología , Técnicas de Cocultivo , Expresión Génica , Granzimas/genética , Granzimas/inmunología , Humanos , Inmunidad Innata , Inmunofenotipificación , Subtipo H7N9 del Virus de la Influenza A/inmunología , Gripe Humana/mortalidad , Gripe Humana/patología , Gripe Humana/virología , Interferón gamma/genética , Interferón gamma/inmunología , Interleucina-18/genética , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/virología , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/virología , Activación de Linfocitos , Monocitos/inmunología , Monocitos/virología , Células T Invariantes Asociadas a Mucosa/virología , Subfamilia B de Receptores Similares a Lectina de Células NK/genética , Subfamilia B de Receptores Similares a Lectina de Células NK/inmunología , Neumonía Viral/mortalidad , Neumonía Viral/patología , Neumonía Viral/virología , Transducción de Señal , Análisis de Supervivencia
13.
J Virol ; 90(15): 6936-6947, 2016 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-27226365

RESUMEN

UNLABELLED: Novel influenza viruses often cause differential infection patterns across different age groups, an effect that is defined as heterogeneous demographic susceptibility. This occurred during the A/H2N2 pandemic, when children experienced higher influenza attack rates than adults. Since the recognition of conserved epitopes across influenza subtypes by CD8(+) cytotoxic T lymphocytes (CTLs) limit influenza disease, we hypothesized that conservation of CTL antigenic peptides (Ag-p) in viruses circulating before the pH2N2-1957 may have resulted in differential CTL immunity. We compared viruses isolated in the years preceding the pandemic (1941 to 1957) to which children and adults were exposed to viruses circulating decades earlier (1918 to 1940), which could infect adults only. Consistent with phylogenetic models, influenza viruses circulating from 1941 to 1957, which infected children, shared with pH2N2 the majority (∼89%) of the CTL peptides within the most immunogenic nucleoprotein, matrix 1, and polymerase basic 1, thus providing evidence for minimal pH2N2 CTL escape in children. Our study, however, identified potential CTL immune evasion from pH2N2 irrespective of age, within HLA-A*03:01(+) individuals for PB1471-L473V/N476I variants and HLA-B*15:01(+) population for NP404-414-V408I mutant. Further experiments using the murine model of B-cell-deficient mice showed that multiple influenza infections resulted in superior protection from influenza-induced morbidity, coinciding with accumulation of tissue-resident memory CD8(+) T cells in the lung. Our study suggests that protection against H2N2-1957 pandemic influenza was most likely linked to the number of influenza virus infections prior to the pandemic challenge rather than differential preexisting CTL immunity. Thus, the regimen of a CTL-based vaccine/vaccine-component may benefit from periodic boosting to achieve fully protective, asymptomatic influenza infection. IMPORTANCE: Due to a lack of cross-reactive neutralizing antibodies, children are particularly susceptible to influenza infections caused by novel viral strains. Preexisting T cell immunity directed at conserved viral regions, however, can provide protection against influenza viruses, promote rapid recovery and better clinical outcomes. When we asked whether high susceptibility of children (compared to adults) to the pandemic H2N2 influenza strain was associated with immune evasion from T-cell immunity, we found high conservation within T-cell antigenic regions in pandemic H2N2. However, the number of influenza infections prior to the challenge was linked to protective, asymptomatic infections and establishment of tissue-resident memory T cells. Our study supports development of vaccines that prime and boost T cells to elicit cross-strain protective T cells, especially tissue-resident memory T cells, for lifelong immunity against distinct influenza viruses.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Epítopos de Linfocito T/inmunología , Gripe Humana/inmunología , Infecciones por Orthomyxoviridae/inmunología , Orthomyxoviridae/inmunología , Pandemias , Linfocitos T Citotóxicos/inmunología , Adulto , Animales , Linfocitos B/inmunología , Niño , Protección Cruzada , Evolución Molecular , Femenino , Humanos , Gripe Humana/virología , Ratones , Infecciones por Orthomyxoviridae/virología , Filogenia
14.
Proc Natl Acad Sci U S A ; 111(2): 769-74, 2014 Jan 14.
Artículo en Inglés | MEDLINE | ID: mdl-24367104

RESUMEN

A unique avian-origin A/H7N9 influenza virus has so far caused 134 cases with 44 deaths. Probing the host factors contributing to disease severity, we found that lower levels of plasma inflammatory cytokines on hospital admission correlated with faster recovery in 18 patients with A/H7N9 influenza virus, whereas high concentrations of (in particular) IL-6, IL-8, and macrophage inflammatory protein-1ß were predictive of a less favorable or fatal outcome. Analysis of bronchoalveolar lavage samples showed up to 1,000-fold greater cytokine/chemokine levels relative to plasma. Furthermore, patients with the rs12252-C/C IFN-induced transmembrane protein-3 (IFITM3) genotype had more rapid disease progression and were less likely to survive. Compared with patients with the rs12252-T/T or rs12252-T/C genotype of IFITM3, patients with the C/C genotype had a shorter time from disease onset to the time point when they sought medical aid (hospital admission or antiviral therapy) and a shorter interval to development of the acute respiratory distress syndrome stage (reflected by shorter intervals between clinical onset and methylprednisolone treatments and higher rates of mechanical ventilator use), as well as experiencing elevated/prolonged lung virus titers and cytokine production and higher mortality. The present analysis provides reported data on the H7N9 influenza-induced "cytokine storm" at the site of infection in humans and identifies the rs12252-C genotype that compromises IFITM3 function as a primary genetic correlate of severe H7N9 pneumonia. Together with rs12252 sequencing, early monitoring of plasma cytokines is thus of prognostic value for the treatment and management of severe influenza pneumonia.


Asunto(s)
Citocinas/inmunología , Brotes de Enfermedades/historia , Subtipo H7N9 del Virus de la Influenza A , Gripe Humana/epidemiología , Gripe Humana/genética , Gripe Humana/inmunología , Proteínas de la Membrana/metabolismo , Proteínas de Unión al ARN/metabolismo , Secuencia de Bases , China/epidemiología , Citocinas/sangre , Cartilla de ADN/genética , Genotipo , Historia del Siglo XXI , Humanos , Pulmón/inmunología , Proteínas de la Membrana/genética , Datos de Secuencia Molecular , Pronóstico , Proteínas de Unión al ARN/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Análisis de Secuencia de ADN , Estadísticas no Paramétricas
15.
J Gen Virol ; 97(12): 3205-3214, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27902386

RESUMEN

FluMist has been used in children and adults for more than 10 years. As pre-existing CD8+ T cell memory pools can provide heterologous immunity against distinct influenza viruses, it is important to understand influenza-specific CD8+ T cell responses elicited by different live attenuated influenza virus (LAIV) regimens. In this study, we immunized mice intranasally with two different doses of live-attenuated PR8 virus (PR8 ts, H1N1), low and high, and then assessed protective efficacy by challenging animals with heterosubtypic X31-H3N2 virus at 6 weeks post-vaccination. Different LAIV doses elicited influenza-specific CD8+ T cell responses in lungs and spleen, but unexpectedly not in bronchoalveolar lavage. Interestingly, the immunodominance hierarchy at the acute phase after immunization varied depending on the LAIV dose; however, these differences disappeared at 6 weeks post-vaccination, resulting in generation of comparable CD8+ T cell memory pools. After vaccination with either dose, sufficient numbers of specific CD8+ T cells were generated for recall and protection of mice against heterosubtypic H1N1→H3N2 challenge. As a result, immunized mice displayed reduced weight loss, diminished inflammatory responses and lower viral titres in lungs, when compared to unvaccinated animals. Interestingly, the higher dose led to enhanced viral clearance on day 5 post-challenge, though this was not associated with increased CD8+ T cell responses, but with higher levels of non-neutralizing antibodies against the priming virus. Our study suggests that, while different LAIV doses result in distinct immune profiles, even a low dose produces sufficient protective CD8+ T cell memory against challenge infection, though the high dose results in more rapid viral clearance and reduced inflammation.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Subtipo H1N1 del Virus de la Influenza A/inmunología , Subtipo H3N2 del Virus de la Influenza A/inmunología , Vacunas contra la Influenza/administración & dosificación , Gripe Humana/inmunología , Vacunas Atenuadas/administración & dosificación , Animales , Anticuerpos Antivirales/inmunología , Humanos , Memoria Inmunológica , Subtipo H1N1 del Virus de la Influenza A/genética , Subtipo H3N2 del Virus de la Influenza A/genética , Vacunas contra la Influenza/inmunología , Gripe Humana/prevención & control , Gripe Humana/virología , Pulmón/virología , Ratones , Ratones Endogámicos C57BL , Vacunas Atenuadas/inmunología
16.
Immunol Cell Biol ; 94(4): 367-77, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26493179

RESUMEN

Indigenous populations, including Indigenous Australians, are highly susceptible to severe influenza disease and the underlying mechanisms are unknown. We studied immune and genetic factors that could predicate severe influenza disease in Indigenous Australians enrolled in the LIFT study: looking into influenza T-cell immunity. To examine CD8(+) T-cell immunity, we characterised human leukocyte antigen (HLA) profiles. HLA typing confirmed previous studies showing predominant usage of HLA-A*02:01, 11:01, 24:02, 34:01 and HLA-B*13:01, 15:21, 40:01/02, 56:01/02 in Indigenous Australians. We identified two new HLA alleles (HLA-A*02:new and HLA-B*56:new). Modelling suggests that variations within HLA-A*02:new (but not HLA-B56:new) could affect peptide binding. There is a relative lack of known influenza epitopes for the majority of these HLAs, with the exception of a universal HLA-A*02:01-M158 epitope and proposed epitopes presented by HLA-A*11:01/HLA-A*24:02. To dissect universal CD8(+) T-cell responses, we analysed the magnitude, function and T-cell receptor (TCR) clonality of HLA-A*02:01-M158(+)CD8(+) T cells. We found comparable IFN-γ, TNF and CD107a and TCRαß characteristics in Indigenous and non-Indigenous Australians, suggesting that the ~15% of Indigenous people that express HLA-A*02:01 have universal influenza-specific CD8(+) T-cell immunity. Furthermore, the frequency of an influenza host risk factor, IFITM3-C/C, was comparable between Indigenous Australians and Europeans, suggesting that expression of this allele does not explain increased disease severity at a population level. Our study indicates a need to identify novel influenza-specific CD8(+) T-cell epitopes restricted by HLA-A and HLA-B alleles prevalent in Indigenous populations for the rational design of universal T-cell vaccines.


Asunto(s)
Linfocitos T CD8-positivos/fisiología , Antígenos HLA/genética , Gripe Humana/inmunología , Nativos de Hawái y Otras Islas del Pacífico , Adulto , Alelos , Células Cultivadas , Epítopos de Linfocito T/metabolismo , Femenino , Prueba de Histocompatibilidad , Humanos , Vacunas contra la Influenza , Gripe Humana/genética , Interferón gamma/metabolismo , Proteína 1 de la Membrana Asociada a los Lisosomas/metabolismo , Masculino , Proteínas de la Membrana/metabolismo , Persona de Mediana Edad , Proteínas de Unión al ARN/metabolismo , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Riesgo , Factor de Necrosis Tumoral alfa/metabolismo , Proteínas de la Matriz Viral/metabolismo
18.
J Virol ; 87(10): 5362-71, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23468508

RESUMEN

Highly pathogenic avian influenza viruses (HPAIV) of subtypes H5 and H7 have caused numerous outbreaks in diverse poultry species and rising numbers of human infections. Both HPAIV subtypes support a growing concern of a pandemic outbreak, specifically via the avian-human link. Natural reassortment of both HPAIV subtypes is a possible event with unpredictable outcome for virulence and host specificity of the progeny virus for avian and mammalian species. NS reassortment of H5N1 HPAIV viruses in the background of A/FPV/Rostock/1934 (H7N1) HPAIV has been shown to change virus replication kinetics and host cell responses in mammalian cells. However, not much is known about the virus-host interaction of such viruses in avian species. In the present study, we show that the NS segment of A/Vietnam/1203/2004 (FPV NS VN, H5N1) HPAIV significantly altered the characteristics of the H7 prototype HPAIV in tracheal organ cultures (TOC) of chicken and turkey in vitro, with decreased replication efficiency accompanied by increased induction of type I interferon (IFN) and apoptosis. Furthermore, species-specific differences between chicken and turkey were demonstrated. Interestingly, NS-reassortant FPV NS VN showed an overall highly pathogenic phenotype, with increased virulence and replication potential compared to the wild-type virus after systemic infection of chicken and turkey embryos. Our data demonstrate that single reassortment of an H5-type NS into an H7-type HPAIV significantly changed virus replication abilities and influenced the avian host cell response without prior adaptation.


Asunto(s)
Interacciones Huésped-Patógeno , Subtipo H5N1 del Virus de la Influenza A/patogenicidad , Subtipo H7N1 del Virus de la Influenza A/patogenicidad , Virus Reordenados/patogenicidad , Proteínas no Estructurales Virales/genética , Animales , Pollos , Subtipo H5N1 del Virus de la Influenza A/genética , Subtipo H5N1 del Virus de la Influenza A/inmunología , Subtipo H5N1 del Virus de la Influenza A/fisiología , Subtipo H7N1 del Virus de la Influenza A/genética , Subtipo H7N1 del Virus de la Influenza A/inmunología , Subtipo H7N1 del Virus de la Influenza A/fisiología , Interferón Tipo I/metabolismo , Técnicas de Cultivo de Órganos , Virus Reordenados/genética , Virus Reordenados/inmunología , Virus Reordenados/fisiología , Tráquea/virología , Pavos , Replicación Viral
19.
Vet Res ; 45: 7, 2014 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-24460592

RESUMEN

Some outbreaks involving highly pathogenic avian influenza viruses (HPAIV) of subtypes H5 and H7 were caused by avian-to-human transmissions. In nature, different influenza A viruses can reassort leading to new viruses with new characteristics. We decided to investigate the impact that the NS-segment of H5 HPAIV would have on viral pathogenicity of a classical avian H7 HPAIV in poultry, a natural host. We focussed this study based on our previous work that demonstrated that single reassortment of the NS-segment from an H5 HPAIV into an H7 HPAIV changes the ability of the virus to replicate in mammalian hosts. Our present data show that two different H7-viruses containing an NS-segment from H5-types (FPV NS GD or FPV NS VN) show an overall highly pathogenic phenotype compared with the wild type H7-virus (FPV), as characterized by higher viral shedding and earlier manifestation of clinical signs. Correlating with the latter, higher amounts of IFN-ß mRNA were detected in the blood of NS-reassortant infected birds, 48 h post-infection (pi). Although lymphopenia was detected in chickens from all AIV-infected groups, also 48 h pi those animals challenged with NS-reassortant viruses showed an increase of peripheral monocyte/macrophage-like cells expressing high levels of IL-1ß, as determined by flow cytometry. Taken together, these findings highlight the importance of the NS-segment in viral pathogenicity which is directly involved in triggering antiviral and pro-inflammatory cytokines found during HPAIV pathogenesis in chickens.


Asunto(s)
Subtipo H5N1 del Virus de la Influenza A/patogenicidad , Subtipo H7N1 del Virus de la Influenza A/patogenicidad , Gripe Aviar/inmunología , Enfermedades de las Aves de Corral/inmunología , Virus Reordenados/patogenicidad , Proteínas no Estructurales Virales/genética , Animales , Pollos , Interacciones Huésped-Patógeno , Subtipo H5N1 del Virus de la Influenza A/genética , Subtipo H5N1 del Virus de la Influenza A/inmunología , Subtipo H5N1 del Virus de la Influenza A/fisiología , Subtipo H7N1 del Virus de la Influenza A/genética , Subtipo H7N1 del Virus de la Influenza A/inmunología , Subtipo H7N1 del Virus de la Influenza A/fisiología , Gripe Aviar/virología , Enfermedades de las Aves de Corral/virología , Virus Reordenados/genética , Virus Reordenados/inmunología , Virus Reordenados/fisiología , Virulencia , Replicación Viral
20.
Expert Rev Vaccines ; 23(1): 362-370, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38444382

RESUMEN

INTRODUCTION: Following the coronavirus disease pandemic, respiratory mucosal vaccines that elicit both mucosal and systemic immune responses have garnered increasing attention. However, human physiological characteristics pose significant challenges in the evaluation of mucosal immunity, which directly impedes the development and application of respiratory mucosal vaccines. AREAS COVERED: This study summarizes the characteristics of immune responses in the respiratory mucosa and reviews the current status and challenges in evaluating immune response to respiratory mucosal vaccines. EXPERT OPINION: Secretory Immunoglobulin A (S-IgA) is a major effector molecule at mucosal sites and a commonly used indicator for evaluating respiratory mucosal vaccines. However, the unique physiological structure of the respiratory tract pose significant challenges for the clinical collection and detection of S-IgA. Therefore, it is imperative to develop a sampling method with high collection efficiency and acceptance, a sensitive detection method, reference materials for mucosal antibodies, and to establish a threshold for S-IgA that correlates with clinical protection. Sample collection is even more challenging when evaluating mucosal cell immunity. Therefore, a mucosal cell sampling method with high operability and high tolerance should be established. Targets of the circulatory system capable of reflecting mucosal cellular immunity should also be explored.


Asunto(s)
Vacunas , Humanos , Inmunidad Mucosa , Inmunoglobulina A Secretora , Mucosa Respiratoria , Vacunación , Anticuerpos Antivirales
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA